Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Cell ; 186(24): 5411-5427.e23, 2023 11 22.
Article in English | MEDLINE | ID: mdl-37918396

ABSTRACT

Neurons build synaptic contacts using different protein combinations that define the specificity, function, and plasticity potential of synapses; however, the diversity of synaptic proteomes remains largely unexplored. We prepared synaptosomes from 7 different transgenic mouse lines with fluorescently labeled presynaptic terminals. Combining microdissection of 5 different brain regions with fluorescent-activated synaptosome sorting (FASS), we isolated and analyzed the proteomes of 18 different synapse types. We discovered ∼1,800 unique synapse-type-enriched proteins and allocated thousands of proteins to different types of synapses (https://syndive.org/). We identify shared synaptic protein modules and highlight the proteomic hotspots for synapse specialization. We reveal unique and common features of the striatal dopaminergic proteome and discover the proteome signatures that relate to the functional properties of different interneuron classes. This study provides a molecular systems-biology analysis of synapses and a framework to integrate proteomic information for synapse subtypes of interest with cellular or circuit-level experiments.


Subject(s)
Brain , Proteome , Synapses , Animals , Mice , Brain/metabolism , Mice, Transgenic , Proteome/metabolism , Proteomics , Synapses/metabolism , Synaptosomes/metabolism
2.
Mol Cell ; 83(11): 1839-1855.e13, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37267905

ABSTRACT

Localized translation is vital to polarized cells and requires precise and robust distribution of different mRNAs and ribosomes across the cell. However, the underlying molecular mechanisms are poorly understood and important players are lacking. Here, we discovered a Rab5 effector, the five-subunit endosomal Rab5 and RNA/ribosome intermediary (FERRY) complex, that recruits mRNAs and ribosomes to early endosomes through direct mRNA-interaction. FERRY displays preferential binding to certain groups of transcripts, including mRNAs encoding mitochondrial proteins. Deletion of FERRY subunits reduces the endosomal localization of transcripts in cells and has a significant impact on mRNA levels. Clinical studies show that genetic disruption of FERRY causes severe brain damage. We found that, in neurons, FERRY co-localizes with mRNA on early endosomes, and mRNA loaded FERRY-positive endosomes are in close proximity of mitochondria. FERRY thus transforms endosomes into mRNA carriers and plays a key role in regulating mRNA distribution and transport.


Subject(s)
Endosomes , rab5 GTP-Binding Proteins , rab5 GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Endosomes/metabolism , Biological Transport , Endocytosis/physiology
3.
J Vis Exp ; (182)2022 04 13.
Article in English | MEDLINE | ID: mdl-35499346

ABSTRACT

Understanding protein homeostasis in vivo is key to knowing how the cells work in both physiological and disease conditions. The present protocol describes in vivo labeling and subsequent purification of newly synthesized proteins using an engineered mouse line to direct protein labeling to specific cellular populations. It is an inducible line by Cre recombinase expression of L274G-Methionine tRNA synthetase (MetRS*), enabling azidonorleucine (ANL) incorporation to the proteins, which otherwise will not occur. Using the method described here, it is possible to purify cell-type-specific proteomes labeled in vivo and detect subtle changes in protein content due to sample complexity reduction.


Subject(s)
Amino Acyl-tRNA Synthetases , Proteome , Amino Acyl-tRNA Synthetases/genetics , Animals , Chromatography, Affinity , Methionine , Mice , Proteostasis
4.
Proc Natl Acad Sci U S A ; 118(43)2021 10 26.
Article in English | MEDLINE | ID: mdl-34670838

ABSTRACT

To form synaptic connections and store information, neurons continuously remodel their proteomes. The impressive length of dendrites and axons imposes logistical challenges to maintain synaptic proteins at locations remote from the transcription source (the nucleus). The discovery of thousands of messenger RNAs (mRNAs) near synapses suggested that neurons overcome distance and gain autonomy by producing proteins locally. It is not generally known, however, if, how, and when localized mRNAs are translated into protein. To investigate the translational landscape in neuronal subregions, we performed simultaneous RNA sequencing (RNA-seq) and ribosome sequencing (Ribo-seq) from microdissected rodent brain slices to identify and quantify the transcriptome and translatome in cell bodies (somata) as well as dendrites and axons (neuropil). Thousands of transcripts were differentially translated between somatic and synaptic regions, with many scaffold and signaling molecules displaying increased translation levels in the neuropil. Most translational changes between compartments could be accounted for by differences in RNA abundance. Pervasive translational regulation was observed in both somata and neuropil influenced by specific mRNA features (e.g., untranslated region [UTR] length, RNA-binding protein [RBP] motifs, and upstream open reading frames [uORFs]). For over 800 mRNAs, the dominant source of translation was the neuropil. We constructed a searchable and interactive database for exploring mRNA transcripts and their translation levels in the somata and neuropil [MPI Brain Research, The mRNA translation landscape in the synaptic neuropil. https://public.brain.mpg.de/dashapps/localseq/ Accessed 5 October 2021]. Overall, our findings emphasize the substantial contribution of local translation to maintaining synaptic protein levels and indicate that on-site translational control is an important mechanism to control synaptic strength.


Subject(s)
Axons/metabolism , Cell Body/metabolism , Dendrites/metabolism , Neurons/metabolism , Protein Biosynthesis , Sequence Analysis, RNA/methods , Animals , Proteome , RNA, Messenger/genetics , Transcriptome
5.
Nat Commun ; 11(1): 4990, 2020 10 05.
Article in English | MEDLINE | ID: mdl-33020478

ABSTRACT

Neurons are highly compartmentalized cells with tightly controlled subcellular protein organization. While brain transcriptome, connectome and global proteome maps are being generated, system-wide analysis of temporal protein dynamics at the subcellular level are currently lacking. Here, we perform a temporally-resolved surfaceome analysis of primary neuron cultures and reveal dynamic surface protein clusters that reflect the functional requirements during distinct stages of neuronal development. Direct comparison of surface and total protein pools during development and homeostatic synaptic scaling demonstrates system-wide proteostasis-independent remodeling of the neuronal surface, illustrating widespread regulation on the level of surface trafficking. Finally, quantitative analysis of the neuronal surface during chemical long-term potentiation (cLTP) reveals fast externalization of diverse classes of surface proteins beyond the AMPA receptor, providing avenues to investigate the requirement of exocytosis for LTP. Our resource (neurosurfaceome.ethz.ch) highlights the importance of subcellular resolution for systems-level understanding of cellular processes.


Subject(s)
Membrane Proteins/metabolism , Neuronal Plasticity , Neurons/metabolism , Synapses/metabolism , Animals , Cell Membrane/metabolism , Cells, Cultured , Excitatory Postsynaptic Potentials , Homeostasis , Long-Term Potentiation , Protein Interaction Maps , Protein Transport , Proteostasis , Rats
6.
Neurobiol Learn Mem ; 173: 107275, 2020 09.
Article in English | MEDLINE | ID: mdl-32659348

ABSTRACT

Long-term memory has been associated with morphological changes in the brain, which in turn tightly correlate with changes in synaptic efficacy. Such plasticity is proposed to rely on dendritic spines as a neuronal canvas on which these changes can occur. Given the key role of actin cytoskeleton dynamics in spine morphology, major regulating factors of this process such as Cofilin 1 (Cfl1) and LIM kinase (LIMK), an inhibitor of Cfl1 activity, are prime molecular targets that may regulate dendritic plasticity. Using a contextual fear conditioning paradigm in mice, we found that pharmacological induction of depolymerization of actin filaments through the inhibition of LIMK causes an impairment in memory reconsolidation, as well as in memory consolidation. On top of that, Cfl1 activity is inhibited and its mRNA is downregulated in CA1 neuropil after re-exposure to the training context. Moreover, by pharmacological disruption of actin cytoskeleton dynamics, the process of memory extinction can either be facilitated or impaired. Our results lead to a better understanding of the role of LIMK, Cfl1 and actin cytoskeleton dynamics in the morphological and functional changes underlying the synaptic plasticity of the memory trace.


Subject(s)
Actins/metabolism , Cofilin 1/metabolism , Fear/physiology , Hippocampus/metabolism , Lim Kinases/metabolism , Memory/physiology , Neuronal Plasticity/physiology , Animals , Male , Memory Consolidation/physiology , Mice
7.
Elife ; 92020 04 24.
Article in English | MEDLINE | ID: mdl-32329716

ABSTRACT

We examined the feedback between the major protein degradation pathway, the ubiquitin-proteasome system (UPS), and protein synthesis in rat and mouse neurons. When protein degradation was inhibited, we observed a coordinate dramatic reduction in nascent protein synthesis in neuronal cell bodies and dendrites. The mechanism for translation inhibition involved the phosphorylation of eIF2α, surprisingly mediated by eIF2α kinase 1, or heme-regulated kinase inhibitor (HRI). Under basal conditions, neuronal expression of HRI is barely detectable. Following proteasome inhibition, HRI protein levels increase owing to stabilization of HRI and enhanced translation, likely via the increased availability of tRNAs for its rare codons. Once expressed, HRI is constitutively active in neurons because endogenous heme levels are so low; HRI activity results in eIF2α phosphorylation and the resulting inhibition of translation. These data demonstrate a novel role for neuronal HRI that senses and responds to compromised function of the proteasome to restore proteostasis.


Subject(s)
Cytoplasm/metabolism , Neurons/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteostasis/physiology , eIF-2 Kinase/metabolism , Animals , Antineoplastic Agents/metabolism , Eukaryotic Initiation Factor-2/metabolism , Heme/metabolism , Mice , Phosphorylation , Rats
8.
Epigenetics Chromatin ; 12(1): 63, 2019 10 10.
Article in English | MEDLINE | ID: mdl-31601272

ABSTRACT

BACKGROUND: MeCP2-a chromatin-binding protein associated with Rett syndrome-has two main isoforms, MeCP2-E1 and MeCP2-E2, differing in a few N-terminal amino acid residues. Previous studies have shown brain region-specific expression of these isoforms which, in addition to their different cellular localization and differential expression during brain development, suggest that they may also have non-overlapping molecular mechanisms. However, differential functions of MeCP2-E1 and E2 remain largely unexplored. RESULTS: Here, we show that the N-terminal domains (NTD) of MeCP2-E1 and E2 modulate the ability of the methyl-binding domain (MBD) to interact with DNA as well as influencing the turn-over rates, binding dynamics, response to neuronal depolarization, and circadian oscillations of the two isoforms. Our proteomics data indicate that both isoforms exhibit unique interacting protein partners. Moreover, genome-wide analysis using ChIP-seq provide evidence for a shared as well as a specific regulation of different sets of genes. CONCLUSIONS: Our study supports the idea that Rett syndrome might arise from simultaneous impairment of cellular processes involving non-overlapping functions of MECP2 isoforms. For instance, MeCP2-E1 mutations might impact stimuli-dependent chromatin regulation, while MeCP2-E2 mutations could result in aberrant ribosomal expression. Overall, our findings provide insight into the functional complexity of MeCP2 by dissecting differential aspects of its two isoforms.


Subject(s)
DNA/metabolism , Methyl-CpG-Binding Protein 2/metabolism , Animals , Cell Line, Tumor , Chromatin/metabolism , Circadian Rhythm/genetics , Humans , Methyl-CpG-Binding Protein 2/chemistry , Methyl-CpG-Binding Protein 2/genetics , Mice , Mice, Knockout , Neurons/metabolism , Protein Binding , Protein Domains , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rats , Rats, Sprague-Dawley , Rett Syndrome/genetics , Rett Syndrome/pathology
9.
J Neurosci ; 39(17): 3188-3203, 2019 04 24.
Article in English | MEDLINE | ID: mdl-30804097

ABSTRACT

Secreted amyloid precursor protein-alpha (sAPPα) has growth factor-like properties and can modulate long-term potentiation (LTP) and memory. Here, we demonstrate that exposure to sAPPα converts short-lasting LTP into protein-synthesis-dependent late LTP in hippocampal slices from male rats. sAPPß had no discernable effect. We hypothesized that sAPPα facilitated LTP via regulated glutamate receptor trafficking and de novo protein synthesis. We found using a linear mixed model that sAPPα stimulated trafficking of GluA2-lacking AMPARs, as well as NMDARs to the extrasynaptic cell surface, in a calcium/calmodulin-dependent kinase II and protein kinase G-dependent manner. Both cell surface receptor accumulation and LTP facilitation were present even after sAPPα washout and inhibition of receptor trafficking or protein synthesis prevented all these effects. Direct visualization of newly synthesized proteins (FUNCAT-PLA) confirmed the ability of sAPPα to stimulate de novo protein synthesis and revealed GluA1 as one of the upregulated proteins. Therefore, sAPPα generates a coordinated synthesis and trafficking of glutamate receptors to the cell surface that facilitate LTP.SIGNIFICANCE STATEMENT Secreted amyloid precursor protein-alpha (sAPPα) is a neurotrophic and neuroprotective protein that can promote synaptic plasticity and memory, yet the molecular mechanisms underlying these effects are still not well understood. Here, we show that sAPPα facilitates long-term potentiation (LTP) in a concentration-dependent fashion through cellular processes involving de novo protein synthesis and trafficking of both GluA2-lacking AMPARs and NMDARs to the extrasynaptic cell surface. sAPPα also enhances GluA1, but not GluA2, synthesis. The trafficking effects, along with the LTP facilitation, persist after sAPPα washout, revealing a metaplastic capability of exogenous sAPPα administration. sAPPα thus facilitates LTP through coordinated activation of protein synthesis and trafficking of glutamate receptors to the cell surface, where they are positioned for priming LTP.


Subject(s)
Amyloid beta-Protein Precursor/pharmacology , Hippocampus/physiology , Long-Term Potentiation/drug effects , Protein Biosynthesis/drug effects , Receptors, Glutamate/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Hippocampus/drug effects , Long-Term Potentiation/physiology , Male , Protein Biosynthesis/physiology , Protein Transport/drug effects , Protein Transport/physiology , Rats , Rats, Sprague-Dawley
10.
Nat Commun ; 10(1): 486, 2019 01 30.
Article in English | MEDLINE | ID: mdl-30700723

ABSTRACT

Drebrin (DBN) regulates cytoskeletal functions during neuronal development, and is thought to contribute to structural and functional synaptic changes associated with aging and Alzheimer's disease. Here we show that DBN coordinates stress signalling with cytoskeletal dynamics, via a mechanism involving kinase ataxia-telangiectasia mutated (ATM). An excess of reactive oxygen species (ROS) stimulates ATM-dependent phosphorylation of DBN at serine-647, which enhances protein stability and accounts for improved stress resilience in dendritic spines. We generated a humanized DBN Caenorhabditis elegans model and show that a phospho-DBN mutant disrupts the protective ATM effect on lifespan under sustained oxidative stress. Our data indicate a master regulatory function of ATM-DBN in integrating cytosolic stress-induced signalling with the dynamics of actin remodelling to provide protection from synapse dysfunction and ROS-triggered reduced lifespan. They further suggest that DBN protein abundance governs actin filament stability to contribute to the consequences of oxidative stress in physiological and pathological conditions.


Subject(s)
Actins/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Oxidative Stress , Actins/genetics , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amino Acid Motifs , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Caenorhabditis elegans , Cells, Cultured , Dendritic Spines/genetics , Dendritic Spines/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropeptides/genetics , Phosphorylation , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
11.
J Neurosci ; 38(29): 6586-6596, 2018 07 18.
Article in English | MEDLINE | ID: mdl-29941446

ABSTRACT

In the nervous system, myelination of axons enables rapid impulse conduction and is a specialized function of glial cells. Myelinating glia are the last cell type to emerge in the evolution of vertebrate nervous systems, presumably in ancient jawed vertebrates (gnathostomata) because jawless vertebrates (agnathans) lack myelin. We have hypothesized that, in these unmyelinated species, evolutionary progenitors of myelinating cells must have existed that should still be present in contemporary agnathan species. Here, we used advanced electron microscopic techniques to reveal axon-glia interactions in the sea lamprey Petromyzon marinus By quantitative assessment of the spinal cord and the peripheral lateral line nerve, we observed a marked maturation-dependent growth of axonal calibers. In peripheral nerves, all axons are ensheathed by glial cells either in bundles or, when larger than the threshold caliber of 3 µm, individually. The ensheathing glia are covered by a basal lamina and express SoxE-transcription factors, features of mammalian Remak-type Schwann cells. In larval lamprey, the ensheathment of peripheral axons leaves gaps that are closed in adults. CNS axons are also covered to a considerable extent by glial processes, which contain a high density of intermediate filaments, glycogen particles, large lipid droplets, and desmosomes, similar to mammalian astrocytes. Indeed, by in situ hybridization, these glial cells express the astrocyte marker Aldh1l1 Specimens were of unknown sex. Our observations imply that radial sorting, ensheathment, and presumably also metabolic support of axons are ancient functions of glial cells that predate the evolutionary emergence of myelin in jawed vertebrates.SIGNIFICANCE STATEMENT We used current electron microscopy techniques to examine axon-glia units in a nonmyelinated vertebrate species, the sea lamprey. In the PNS, lamprey axons are fully ensheathed either individually or in bundles by cells ortholog to Schwann cells. In the CNS, axons associate with astrocyte orthologs, which contain glycogen and lipid droplets. We suggest that ensheathment, radial sorting, and metabolic support of axons by glial cells predate the evolutionary emergence of myelin in ancient jawed vertebrates.


Subject(s)
Axons/metabolism , Axons/ultrastructure , Myelin Sheath/metabolism , Myelin Sheath/ultrastructure , Neuroglia/metabolism , Animals , Biological Evolution , Lampreys , Neurogenesis/physiology
12.
Nat Biotechnol ; 35(12): 1196-1201, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29106408

ABSTRACT

Although advances in protein labeling methods have made it possible to measure the proteome of mixed cell populations, it has not been possible to isolate cell-type-specific proteomes in vivo. This is because the existing methods for metabolic protein labeling in vivo access all cell types. We report the development of a transgenic mouse line where Cre-recombinase-induced expression of a mutant methionyl-tRNA synthetase (L274G) enables the cell-type-specific labeling of nascent proteins with a non-canonical amino-acid and click chemistry. Using immunoblotting, imaging and mass spectrometry, we use our transgenic mouse to label and analyze proteins in excitatory principal neurons and Purkinje neurons in vitro (brain slices) and in vivo. We discover more than 200 proteins that are differentially regulated in hippocampal excitatory neurons by exposing mice to an environment with enriched sensory cues. Our approach can be used to isolate, analyze and quantitate cell-type-specific proteomes and their dynamics in healthy and diseased tissues.


Subject(s)
Gene Expression Regulation/genetics , Proteome/genetics , Proteome/metabolism , Proteomics/methods , Amino Acids/analysis , Amino Acids/chemistry , Amino Acids/metabolism , Animals , Click Chemistry , Female , Gene Expression Regulation/physiology , Integrases/genetics , Integrases/metabolism , Male , Methionine-tRNA Ligase/metabolism , Mice , Mice, Transgenic , Neurons/chemistry , Neurons/metabolism , Proteome/analysis , Proteome/chemistry
13.
EMBO Rep ; 18(5): 693-711, 2017 05.
Article in English | MEDLINE | ID: mdl-28404606

ABSTRACT

Efficient neuronal function depends on the continued modulation of the local neuronal proteome. Local protein synthesis plays a central role in tuning the neuronal proteome at specific neuronal regions. Various aspects of translation such as the localization of translational machinery, spatial spread of the newly translated proteins, and their site of action are carried out in specialized neuronal subcompartments to result in a localized functional outcome. In this review, we focus on the various aspects of these local translation compartments such as size, biochemical and organelle composition, structural boundaries, and temporal dynamics. We also discuss the apparent absence of definitive components of translation in these local compartments and the emerging state-of-the-art tools that could help dissecting these conundrums in greater detail in the future.


Subject(s)
Neuronal Plasticity , Neurons/physiology , Protein Biosynthesis , Animals , Dendrites/physiology , Neurons/cytology , Organelles/physiology , Proteome , RNA, Messenger/genetics , RNA, Messenger/metabolism
14.
Nat Methods ; 12(5): 411-4, 2015 May.
Article in English | MEDLINE | ID: mdl-25775042

ABSTRACT

Protein synthesis is a dynamic process that tunes the cellular proteome in response to internal and external demands. Metabolic labeling approaches identify the general proteomic response but cannot visualize specific newly synthesized proteins within cells. Here we describe a technique that couples noncanonical amino acid tagging or puromycylation with the proximity ligation assay to visualize specific newly synthesized proteins and monitor their origin, redistribution and turnover in situ.


Subject(s)
Fibroblasts/metabolism , Proteins/chemistry , Proteins/metabolism , Animals , Antibodies , Cells, Cultured , Gene Expression Regulation/physiology , Hippocampus/cytology , Mice , Neurons/metabolism , Rats , Staining and Labeling
15.
Angew Chem Int Ed Engl ; 54(12): 3717-21, 2015 Mar 16.
Article in English | MEDLINE | ID: mdl-25656536

ABSTRACT

The antibiotic puromycin, which inhibits protein translation, is used in a broad range of biochemical applications. The synthesis, characterization, and biological applications of NVOC-puromycin, a photocaged derivative that is activated by UV illumination, are presented. The caged compound had no effect either on prokaryotic or eukaryotic translation or on the viability of HEK 293 cells. Furthermore, no significant release of ribosome-bound polypeptide chains was detected in vitro. Upon illumination, cytotoxic activity, in vitro translation inhibition, and polypeptide release triggered by the uncaging of NVOC-puromycin were equivalent to those of the commercial compound. The quantum yield of photolysis was determined to be 1.1±0.2% and the NVOC-puromycin was applied to the detection of newly translated proteins with remarkable spatiotemporal resolution by using two-photon laser excitation, puromycin immunohistochemistry, and imaging in rat hippocampal neurons.


Subject(s)
Peptides/chemistry , Puromycin/chemistry , Animals , Benzaldehydes/chemistry , Cell Survival/drug effects , HEK293 Cells , Hippocampus/metabolism , Humans , Microscopy, Fluorescence , Peptides/metabolism , Photolysis/radiation effects , Protein Biosynthesis/drug effects , Puromycin/toxicity , Rats , Ultraviolet Rays
16.
Cell Rep ; 7(6): 1771-8, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24931613

ABSTRACT

Localized signaling in neuronal dendrites requires tight spatial control of membrane composition. Upon initial synthesis, nascent secretory cargo in dendrites exits the endoplasmic reticulum (ER) from local zones of ER complexity that are spatially coupled to post-ER compartments. Although newly synthesized membrane proteins can be processed locally, the mechanisms that control the spatial range of secretory cargo transport in dendritic segments are unknown. Here, we monitored the dynamics of nascent membrane proteins in dendritic post-ER compartments under regimes of low or increased neuronal activity. In response to activity blockade, post-ER carriers are highly mobile and are transported over long distances. Conversely, increasing synaptic activity dramatically restricts the spatial scale of post-ER trafficking along dendrites. This activity-induced confinement of secretory cargo requires site-specific phosphorylation of the kinesin motor KIF17 by Ca(2+)/calmodulin-dependent protein kinases (CaMK). Thus, the length scales of early secretory trafficking in dendrites are tuned by activity-dependent regulation of microtubule-dependent transport.


Subject(s)
Dendrites/metabolism , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Synapses/metabolism , Animals , Biological Transport , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Kinesins/metabolism , Molecular Motor Proteins/metabolism , Phosphorylation , Protein Transport
18.
Sci Signal ; 6(306): rs16, 2013 Dec 17.
Article in English | MEDLINE | ID: mdl-24345682

ABSTRACT

Brain-derived neurotrophic factor (BDNF) is a small protein of the neurotrophin family that regulates various brain functions. Although much is known about how its transcription is regulated, the abundance of endogenous BDNF mRNA and its subcellular localization pattern are matters of debate. We used next-generation sequencing and high-resolution in situ hybridization in the rat hippocampus to reexamine this question. We performed 3' end sequencing on rat hippocampal slices and detected two isoforms of Bdnf containing either a short or a long 3' untranslated region (3'UTR). Most of the Bdnf transcripts contained the short 3'UTR isoform and were present in low amounts relative to other neuronal transcripts. Bdnf mRNA was present in the somatic compartment of rat hippocampal slices or the somata of cultured rat hippocampal neurons but was rarely detected in the dendritic processes. Pharmacological stimulation of hippocampal neurons induced Bdnf expression but did not change the ratio of Bdnf isoform abundance. The findings indicate that endogenous Bdnf mRNA, although weakly abundant, is primarily localized to the somatic compartment of hippocampal neurons. Both Bdnf mRNA isoforms have shorter half-lives compared with other neuronal mRNAs. Furthermore, the findings show that using complementary high-resolution techniques can provide sensitive measures of endogenous transcript abundance.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , High-Throughput Nucleotide Sequencing , Hippocampus/metabolism , Neurons/metabolism , RNA, Messenger/genetics , 3' Untranslated Regions , Animals , Hippocampus/cytology , In Situ Hybridization , Rats
19.
Channels (Austin) ; 7(6): 418-9, 2013.
Article in English | MEDLINE | ID: mdl-24722264

ABSTRACT

How we see our environment is the result of a multi-level, parallel processing effort by the central nervous system. This computation is initiated within the retina at the very first synapse in the visual pathway - the photoreceptor ribbon synapse. Two recent studies shed light on the critical role of balanced calcium channel activity in maturation of this highly specialized synapse. (1, 2.)


Subject(s)
Calcium Channels/genetics , Calcium Channels/metabolism , Eye Diseases, Hereditary/genetics , Eye Diseases, Hereditary/metabolism , Eye Diseases, Hereditary/pathology , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/metabolism , Genetic Diseases, X-Linked/pathology , Myopia/genetics , Myopia/metabolism , Myopia/pathology , Night Blindness/genetics , Night Blindness/metabolism , Night Blindness/pathology , Retinal Cone Photoreceptor Cells/pathology , Retinal Rod Photoreceptor Cells/pathology , Synapses/metabolism , Animals , Calcium Channels, L-Type , Female , Humans , Male
20.
Curr Protoc Cell Biol ; Chapter 7: 7.11.1-7.11.29, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22968844

ABSTRACT

Fluorescent labeling of proteins by genetically encoded fluorescent protein tags has enabled an enhanced understanding of cell biological processes but is restricted to the analysis of a limited number of identified proteins. This approach does not permit, e.g., the unbiased visualization of a full proteome in situ. We describe here a fluorescence-based method to follow proteome-wide patterns of newly synthesized proteins in cultured cells, tissue slices, and a whole organism. This technique is compatible with immunohistochemistry and in situ hybridization. Key to this method is the introduction of a small bio-orthogonal reactive group by metabolic labeling. This is accomplished by replacing the amino acid methionine by the azide-bearing methionine surrogate azidohomoalanine (AHA) in a step very similar to classical radioisotope labeling. Subsequently, an alkyne-bearing fluorophore is covalently attached to the group by "click chemistry"--a copper(I)-catalyzed [3+2]azide-alkyne cycloaddition. By similar means, metabolic labeling can also be performed with the alkyne-bearing homopropargylglycine (HPG) and clicked to an azide-functionalized fluorophore.


Subject(s)
Azides/chemistry , Cell Culture Techniques/methods , Fluorescent Dyes/chemistry , Immunohistochemistry/methods , In Situ Hybridization, Fluorescence/methods , Methionine/metabolism , Proteins/chemistry , Animals , Azides/metabolism , Fluorescent Dyes/metabolism , Humans , Methionine/chemistry , Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...